Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Nat Commun ; 13(1): 4188, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35858913

ABSTRACT

The formation of a functional blood vessel network relies on the ability of endothelial cells (ECs) to dynamically rearrange their adhesive contacts in response to blood flow and guidance cues, such as vascular endothelial growth factor-A (VEGF-A) and class 3 semaphorins (SEMA3s). Neuropilin 1 (NRP1) is essential for blood vessel development, independently of its ligands VEGF-A and SEMA3, through poorly understood mechanisms. Grounding on unbiased proteomic analysis, we report here that NRP1 acts as an endocytic chaperone primarily for adhesion receptors on the surface of unstimulated ECs. NRP1 localizes at adherens junctions (AJs) where, interacting with VE-cadherin, promotes its basal internalization-dependent turnover and favors vascular permeability initiated by histamine in both cultured ECs and mice. We identify a splice variant of tryptophanyl-tRNA synthetase (mini-WARS) as an unconventionally secreted extracellular inhibitory ligand of NRP1 that, by stabilizing it at the AJs, slows down both VE-cadherin turnover and histamine-elicited endothelial leakage. Thus, our work shows a role for NRP1 as a major regulator of AJs plasticity and reveals how mini-WARS acts as a physiological NRP1 inhibitory ligand in the control of VE-cadherin endocytic turnover and vascular permeability.


Subject(s)
Neuropilin-1 , Tryptophan-tRNA Ligase , Adherens Junctions/metabolism , Animals , Antigens, CD , Cadherins/genetics , Capillary Permeability , Endothelial Cells/metabolism , Histamine , Ligands , Mice , Neuropilin-1/genetics , Neuropilin-1/metabolism , Proteomics , Tryptophan-tRNA Ligase/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
Cancer Immunol Res ; 9(3): 309-323, 2021 03.
Article in English | MEDLINE | ID: mdl-33361087

ABSTRACT

IL1ß is a central mediator of inflammation. Secretion of IL1ß typically requires proteolytic maturation by the inflammasome and formation of membrane pores by gasdermin D (GSDMD). Emerging evidence suggests an important role for IL1ß in promoting cancer progression in patients, but the underlying mechanisms are ill-defined. Here, we have shown a key role for IL1ß in driving tumor progression in two distinct mouse tumor models. Notably, activation of the inflammasome, caspase-8, as well as the pore-forming proteins GSDMD and mixed lineage kinase domain-like protein in the host were dispensable for the release of intratumoral bioactive IL1ß. Inflammasome-independent IL1ß release promoted systemic neutrophil expansion and fostered accumulation of T-cell-suppressive neutrophils in the tumor. Moreover, IL1ß was essential for neutrophil infiltration triggered by antiangiogenic therapy, thereby contributing to treatment-induced immunosuppression. Deletion of IL1ß allowed intratumoral accumulation of CD8+ effector T cells that subsequently activated tumor-associated macrophages. Depletion of either CD8+ T cells or macrophages abolished tumor growth inhibition in IL1ß-deficient mice, demonstrating a crucial role for CD8+ T-cell-macrophage cross-talk in the antitumor immune response. Overall, these results support a tumor-promoting role for IL1ß through establishing an immunosuppressive microenvironment and show that inflammasome activation is not essential for release of this cytokine in tumors.


Subject(s)
Interleukin-1beta/metabolism , Neoplasms/immunology , Neutrophils/immunology , Tumor Escape , Tumor Microenvironment/immunology , Animals , Cell Communication/immunology , Disease Models, Animal , Female , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Knockout , Neoplasms/pathology , Neutrophils/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Tumor-Associated Macrophages/immunology
3.
EMBO Mol Med ; 12(10): e11210, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32885605

ABSTRACT

Glutamine synthetase (GS) generates glutamine from glutamate and controls the release of inflammatory mediators. In macrophages, GS activity, driven by IL10, associates to the acquisition of M2-like functions. Conditional deletion of GS in macrophages inhibits metastasis by boosting the formation of anti-tumor, M1-like, tumor-associated macrophages (TAMs). From this basis, we evaluated the pharmacological potential of GS inhibitors in targeting metastasis, identifying glufosinate as a specific human GS inhibitor. Glufosinate was tested in both cultured macrophages and on mice bearing metastatic lung, skin and breast cancer. We found that glufosinate rewires macrophages toward an M1-like phenotype both at the primary tumor and metastatic site, countering immunosuppression and promoting vessel sprouting. This was also accompanied to a reduction in cancer cell intravasation and extravasation, leading to synchronous and metachronous metastasis growth inhibition, but no effects on primary tumor growth. Glufosinate treatment was well-tolerated, without liver and brain toxicity, nor hematopoietic defects. These results identify GS as a druggable enzyme to rewire macrophage functions and highlight the potential of targeting metabolic checkpoints in macrophages to treat cancer metastasis.


Subject(s)
Breast Neoplasms , Macrophages , Aminobutyrates , Animals , Female , Humans , Inflammation Mediators , Mice
4.
Circ Res ; 127(6): 707-723, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32527198

ABSTRACT

RATIONALE: How endothelial cells (ECs) migrate and form an immature vascular plexus has been extensively studied. Yet, mechanisms underlying vascular remodeling remain poorly established. A better understanding of these processes may lead to the design of novel therapeutic strategies complementary to current angiogenesis inhibitors. OBJECTIVE: Starting from our previous observations that PP2A (protein phosphatase 2) regulates the HIF (hypoxia-inducible factor)/PHD-2 (prolyl hydroxylase 2)-constituted oxygen machinery, we hypothesized that this axis could play an important role during blood vessel formation, tissue perfusion, and oxygen restoration. METHODS AND RESULTS: We show that the PP2A regulatory subunit B55α is at the crossroad between vessel pruning and vessel maturation. Blood vessels with high B55α counter cell stress conditions and thrive for stabilization and maturation. When B55α is inhibited, ECs cannot cope with cell stress and undergo apoptosis, leading to massive pruning of nascent blood vessels. Mechanistically, we found that the B55α/PP2A complex restrains PHD-2 activity, promoting EC survival in a HIF-dependent manner, and furthermore dephosphorylates p38, altogether protecting ECs against cell stress occurring, for example, during the onset of blood flow. In tumors, EC-specific B55α deficiency induces pruning of immature-like tumor blood vessels resulting in delayed tumor growth and metastasis, without affecting nonpathological vessels. Consistently, systemic administration of a pan-PP2A inhibitor disrupts vascular network formation and tumor progression in vivo without additional effects on B55α-deficient vessels. CONCLUSIONS: Our data underline a unique role of the B55α/PP2A phosphatase complex in vessel remodeling and suggest the use of PP2A-inhibitors as potent antiangiogenic drugs targeting specifically nascent blood vessels with a mode-of-action complementary to VEGF-R (vascular endothelial growth factor receptor)-targeted therapies. Graphical Abstract: A graphical abstract is available for this article.


Subject(s)
Apoptosis , Breast Neoplasms/enzymology , Carcinoma, Lewis Lung/enzymology , Endothelial Cells/enzymology , Neovascularization, Pathologic , Protein Phosphatase 2/metabolism , Vascular Remodeling , Angiogenesis Inhibitors/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Inhibitors/pharmacology , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Cell Metab ; 30(5): 917-936.e10, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31447322

ABSTRACT

Among mammary tumor-infiltrating immune cells, the highest expression of podoplanin (PDPN) is found in a subset of tumor-associated macrophages (TAMs). We hereby demonstrate that PDPN is involved in the attachment of this TAM subset to lymphatic endothelial cells (LECs). Mechanistically, the binding of PDPN to LEC-derived galectin 8 (GAL8) in a glycosylation-dependent manner promotes the activation of pro-migratory integrin ß1. When proximal to lymphatics, PDPN-expressing macrophages (PoEMs) stimulate local matrix remodeling and promote vessel growth and lymphoinvasion. Anti-integrin ß1 blockade, macrophage-specific Pdpn knockout, or GAL8 inhibition impairs TAM adhesion to LECs, restraining lymphangiogenesis and reducing lymphatic cancer spread. In breast cancer patients, association of PoEMs with tumor lymphatic vessels correlates with incidences of lymph node and distant organ metastasis.


Subject(s)
Breast Neoplasms/metabolism , Lymph Nodes/pathology , Lymphangiogenesis/genetics , Lymphatic Metastasis/genetics , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Adult , Aged , Aged, 80 and over , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Female , Humans , Lymphatic Vessels/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged
6.
Cell Metab ; 30(1): 16-18, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31269424

ABSTRACT

Cancer cells are highly heterogeneous, and their features markedly vary within different areas of the tumor microenvironment. In this issue, Kumar et al. (2019) identified perivascular tumor cells, derived from mouse glioblastoma xenografts, as the fraction that displays the highest mTOR-dependent anabolic metabolism, aggressiveness, and resistance to therapy.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Heterografts , Mice , Transplantation, Heterologous , Tumor Microenvironment
7.
Arterioscler Thromb Vasc Biol ; 38(4): 854-869, 2018 04.
Article in English | MEDLINE | ID: mdl-29449332

ABSTRACT

OBJECTIVE: Endothelial upregulation of adhesion molecules serves to recruit leukocytes to inflammatory sites and appears to be promoted by NOTCH1; however, current models based on interactions between active NOTCH1 and NF-κB components cannot explain the transcriptional selectivity exerted by NOTCH1 in this context. APPROACH AND RESULTS: Observing that Cre/Lox-induced conditional mutations of endothelial Notch modulated inflammation in murine contact hypersensitivity, we found that IL (interleukin)-1ß stimulation induced rapid recruitment of RELA (v-rel avian reticuloendotheliosis viral oncogene homolog A) to genomic sites occupied by NOTCH1-RBPJ (recombination signal-binding protein for immunoglobulin kappa J region) and that NOTCH1 knockdown reduced histone H3K27 acetylation at a subset of NF-κB-directed inflammatory enhancers. CONCLUSIONS: Our findings reveal that NOTCH1 signaling supports the expression of a subset of inflammatory genes at the enhancer level and demonstrate how key signaling pathways converge on chromatin to coordinate the transition to an infla mmatory endothelial phenotype.


Subject(s)
Endothelial Cells/drug effects , Histones/metabolism , Inflammation/prevention & control , Interleukin-1beta/pharmacology , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Acetylation , Animals , Appendicitis/metabolism , Appendicitis/pathology , Cells, Cultured , Dermatitis, Contact/genetics , Dermatitis, Contact/metabolism , Dermatitis, Contact/pathology , Dipeptides/pharmacology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptor, Notch1/genetics , Signal Transduction/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
8.
Cell Rep ; 21(10): 2842-2854, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29212030

ABSTRACT

Although it is well established that tumor-associated macrophages take part in each step of cancer progression, less is known about the distinct role of the so-called metastasis-associated macrophages (MAMs) at the metastatic site. Previous studies reported that Caveolin-1 (Cav1) has both tumor-promoting and tumor-suppressive functions. However, the role of Cav1 in bone-marrow-derived cells is unknown. Here, we describe Cav1 as an anti-metastatic regulator in mouse models of lung and breast cancer pulmonary metastasis. Among all the recruited inflammatory cell populations, we show that MAMs uniquely express abundant levels of Cav1. Using clodronate depletion of macrophages, we demonstrate that macrophage Cav1 signaling is critical for metastasis and not for primary tumor growth. In particular, Cav1 inhibition does not affect MAM recruitment to the metastatic site but, in turn, favors angiogenesis. We describe a mechanism by which Cav1 in MAMs specifically restrains vascular endothelial growth factor A/vascular endothelial growth factor receptor 1 (VEGF-A/VEGFR1) signaling and its downstream effectors, matrix metallopeptidase 9 (MMP9) and colony-stimulating factor 1 (CSF1).


Subject(s)
Caveolin 1/metabolism , Macrophages/metabolism , Animals , Caveolin 1/genetics , Lung Neoplasms/metabolism , Mice , Mice, Knockout , Neoplasm Metastasis , Neovascularization, Pathologic/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Nat Commun ; 8(1): 2210, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263363

ABSTRACT

VEGFR-2/Notch signalling regulates angiogenesis in part by driving the remodelling of endothelial cell junctions and by inducing cell migration. Here, we show that VEGF-induced polarized cell elongation increases cell perimeter and decreases the relative VE-cadherin concentration at junctions, triggering polarized formation of actin-driven junction-associated intermittent lamellipodia (JAIL) under control of the WASP/WAVE/ARP2/3 complex. JAIL allow formation of new VE-cadherin adhesion sites that are critical for cell migration and monolayer integrity. Whereas at the leading edge of the cell, large JAIL drive cell migration with supportive contraction, lateral junctions show small JAIL that allow relative cell movement. VEGFR-2 activation initiates cell elongation through dephosphorylation of junctional myosin light chain II, which leads to a local loss of tension to induce JAIL-mediated junctional remodelling. These events require both microtubules and polarized Rac activity. Together, we propose a model where polarized JAIL formation drives directed cell migration and junctional remodelling during sprouting angiogenesis.


Subject(s)
Actins/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Movement/physiology , Cell Polarity/physiology , Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/metabolism , Actin-Related Protein 2/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 3/metabolism , Actins/drug effects , Antigens, CD/drug effects , Cadherins/drug effects , Cardiac Myosins/metabolism , Cell Adhesion , Cell Movement/drug effects , Cell Polarity/drug effects , Endothelial Cells/drug effects , Endothelial Cells/physiology , Endothelium, Vascular , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions/drug effects , Microtubules/drug effects , Microtubules/metabolism , Models, Cardiovascular , Myosin Light Chains/metabolism , Neovascularization, Physiologic/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism , Pseudopodia/physiology , Signal Transduction , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Remodeling , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac GTP-Binding Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 114(40): 10725-10730, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28923935

ABSTRACT

Retinoid X receptor (RXR) regulates several key functions in myeloid cells, including inflammatory responses, phagocytosis, chemokine secretion, and proangiogenic activity. Its importance, however, in tumor-associated myeloid cells is unknown. In this study, we demonstrate that deletion of RXR in myeloid cells enhances lung metastasis formation while not affecting primary tumor growth. We show that RXR deficiency leads to transcriptomic changes in the lung myeloid compartment characterized by increased expression of prometastatic genes, including important determinants of premetastatic niche formation. Accordingly, RXR-deficient myeloid cells are more efficient in promoting cancer cell migration and invasion. Our results suggest that the repressive activity of RXR on prometastatic genes is mediated primarily through direct DNA binding of the receptor along with nuclear receptor corepressor (NCoR) and silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) corepressors and is largely unresponsive to ligand activation. In addition, we found that expression and transcriptional activity of RXRα is down-modulated in peripheral blood mononuclear cells of patients with lung cancer, particularly in advanced and metastatic disease. Overall, our results identify RXR as a regulator in the myeloid cell-assisted metastatic process and establish lipid-sensing nuclear receptors in the microenvironmental regulation of tumor progression.


Subject(s)
Carcinoma, Lewis Lung/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Myeloid Cells/pathology , Retinoid X Receptors/physiology , Transcription, Genetic , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Cells, Cultured , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism
11.
Nat Cell Biol ; 19(8): 915-927, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28714968

ABSTRACT

Endothelial sprouting and proliferation are tightly coordinated processes mediating the formation of new blood vessels during physiological and pathological angiogenesis. Endothelial tip cells lead sprouts and are thought to suppress tip-like behaviour in adjacent stalk endothelial cells by activating Notch. Here, we show with genetic experiments in postnatal mice that the level of active Notch signalling is more important than the direct Dll4-mediated cell-cell communication between endothelial cells. We identify endothelial expression of VEGF-A and of the chemokine receptor CXCR4 as key processes controlling Notch-dependent vessel growth. Surprisingly, genetic experiments targeting endothelial tip cells in vivo reveal that they retain their function without Dll4 and are also not replaced by adjacent, Dll4-positive cells. Instead, activation of Notch directs tip-derived endothelial cells into developing arteries and thereby establishes that Dll4-Notch signalling couples sprouting angiogenesis and artery formation.


Subject(s)
Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Receptor, Notch1/metabolism , Retinal Artery/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell Communication , Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Cells, Cultured , Female , Gene Expression Regulation , Genotype , Intracellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptor, Notch1/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Retinal Artery/cytology , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Cell Rep ; 18(7): 1699-1712, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28199842

ABSTRACT

Oxygen-dependent HIF1α hydroxylation and degradation are strictly controlled by PHD2. In hypoxia, HIF1α partly escapes degradation because of low oxygen availability. Here, we show that PHD2 is phosphorylated on serine 125 (S125) by the mechanistic target of rapamycin (mTOR) downstream kinase P70S6K and that this phosphorylation increases its ability to degrade HIF1α. mTOR blockade in hypoxia by REDD1 restrains P70S6K and unleashes PP2A phosphatase activity. Through its regulatory subunit B55α, PP2A directly dephosphorylates PHD2 on S125, resulting in a further reduction of PHD2 activity that ultimately boosts HIF1α accumulation. These events promote autophagy-mediated cell survival in colorectal cancer (CRC) cells. B55α knockdown blocks neoplastic growth of CRC cells in vitro and in vivo in a PHD2-dependent manner. In patients, CRC tissue expresses higher levels of REDD1, B55α, and HIF1α but has lower phospho-S125 PHD2 compared with a healthy colon. Our data disclose a mechanism of PHD2 regulation that involves the mTOR and PP2A pathways and controls tumor growth.


Subject(s)
Cell Hypoxia/physiology , Cell Survival/physiology , Colorectal Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Protein Phosphatase 2/metabolism , TOR Serine-Threonine Kinases/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , HEK293 Cells , HT29 Cells , Humans , Phosphorylation/physiology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/physiology
13.
Trends Mol Med ; 22(2): 85-87, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26774932

ABSTRACT

Targeting the aberrant, nonfunctional tumor vasculature is one of the most promising approaches of new anticancer therapies. Here, we discuss a recent publication and a novel mechanism by which tumor-associated macrophages can actively induce vessel normalization and junction stability, and improve the distribution of therapeutic drugs into a tumor.


Subject(s)
Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Humans
14.
Nat Commun ; 6: 6429, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25752958

ABSTRACT

Angiogenic blood vessel growth requires several distinct but integrated cellular activities. Endothelial cell sprouting and proliferation lead to the expansion of the vasculature and give rise to a highly branched, immature plexus, which is subsequently reorganized into a mature and stable network. Although it is known that integrin-mediated cell-matrix interactions are indispensable for embryonic angiogenesis, little is known about the function of integrins in different steps of vascular morphogenesis. Here, by investigating the integrin ß1-subunit with inducible and endothelial-specific gene targeting in the postnatal mouse retina, we show that ß1 integrin promotes endothelial sprouting but is a negative regulator of proliferation. In maturing vessels, integrin ß1 is indispensable for proper localization of VE-cadherin and thereby cell-cell junction integrity. The sum of our findings establishes that integrin ß1 has critical functions in the growing and maturing vasculature, and is required for the formation of stable, non-leaky blood vessels.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Endothelium/growth & development , Integrin beta1/metabolism , Intercellular Junctions/physiology , Morphogenesis/physiology , Neovascularization, Physiologic/physiology , Retinal Vessels/growth & development , Animals , Blotting, Western , Brain/anatomy & histology , Cell Proliferation , Endothelium/metabolism , Gene Targeting , Image Processing, Computer-Assisted , Immunohistochemistry , Immunoprecipitation , Intercellular Junctions/metabolism , Mice , Microscopy, Electron , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Retinal Vessels/ultrastructure
15.
J Exp Med ; 211(2): 281-95, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24446488

ABSTRACT

Cytochrome P450 (CYP) epoxygenases generate bioactive lipid epoxides which can be further metabolized to supposedly less active diols by the soluble epoxide hydrolase (sEH). As the role of epoxides and diols in angiogenesis is unclear, we compared retinal vasculature development in wild-type and sEH(-/-) mice. Deletion of the sEH significantly delayed angiogenesis, tip cell, and filopodia formation, a phenomenon associated with activation of the Notch signaling pathway. In the retina, sEH was localized in Müller glia cells, and Müller cell-specific sEH deletion reproduced the sEH(-/-) retinal phenotype. Lipid profiling revealed that sEH deletion decreased retinal and Müller cell levels of 19,20-dihydroxydocosapentaenoic acid (DHDP), a diol of docosahexenoic acid (DHA). 19,20-DHDP suppressed endothelial Notch signaling in vitro via inhibition of the γ-secretase and the redistribution of presenilin 1 from lipid rafts. Moreover, 19,20-DHDP, but not the parent epoxide, was able to rescue the defective angiogenesis in sEH(-/-) mice as well as in animals lacking the Fbxw7 ubiquitin ligase, which demonstrate strong basal activity of the Notch signaling cascade. These studies demonstrate that retinal angiogenesis is regulated by a novel form of neuroretina-vascular interaction involving the sEH-dependent generation of a diol of DHA in Müller cells.


Subject(s)
Ependymoglial Cells/metabolism , Epoxide Hydrolases/metabolism , Fatty Acids, Unsaturated/biosynthesis , Neovascularization, Physiologic , Receptors, Notch/metabolism , Retinal Vessels/growth & development , Retinal Vessels/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Astrocytes/metabolism , Epoxide Hydrolases/deficiency , Epoxide Hydrolases/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinal Vessels/cytology , Signal Transduction , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
16.
Development ; 140(14): 3051-61, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23785053

ABSTRACT

Blood vessels form a hierarchically organized network of arteries, capillaries and veins, which develops through a series of growth, pruning and maturation processes. In contrast to the rapidly increasing insight into the processes controlling vascular growth and, in particular, endothelial sprouting and proliferation, the conversion of immature vessels into a fully functional, quiescent vasculature remains little understood. Here we used inducible, cell type-specific genetic approaches to show that endothelial Notch signaling is crucial for the remodeling of veins and the perivenous capillary plexus, which occurs after the completion of the initial angiogenic growth phase in the retina of adolescent mice. Mutant vessels showed ectopic proliferation and sprouting, defective recruitment of supporting mural cells, and failed to downregulate the expression of VEGF receptors. Surprisingly, by contrast Notch was dispensable in the endothelium of remodeling postnatal arteries. Taken together, our results identify key processes contributing to vessel remodeling, maturation and the acquisition of a quiescent phenotype in the final stage of developmental angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic , Receptors, Notch/metabolism , Retinal Vessels/cytology , Signal Transduction , Animals , Cell Proliferation , Down-Regulation , Gene Targeting , Mice , Receptors, Vascular Endothelial Growth Factor/genetics , Retinal Artery/cytology , Retinal Vessels/growth & development , Retinal Vessels/metabolism
17.
PLoS One ; 7(7): e41116, 2012.
Article in English | MEDLINE | ID: mdl-22848434

ABSTRACT

Notch signaling controls fundamental aspects of angiogenic blood vessel growth including the selection of sprouting tip cells, endothelial proliferation and arterial differentiation. The E3 ubiquitin ligase Fbxw7 is part of the SCF protein complex responsible for the polyubiquitination and thereby proteasomal degradation of substrates such as Notch, c-Myc and c-Jun. Here, we show that Fbxw7 is a critical regulator of angiogenesis in the mouse retina and the zebrafish embryonic trunk, which we attribute to its role in the degradation of active Notch. Growth of retinal blood vessel was impaired and the Notch ligand Dll4, which is also a Notch target, upregulated in inducible and endothelial cell-specific Fbxw7(iECKO) mutant mice. The stability of the cleaved and active Notch intracellular domain was increased after siRNA knockdown of the E3 ligase in cultured human endothelial cells. Injection of fbxw7 morpholinos interfered with the sprouting of zebrafish intersegmental vessels (ISVs). Arguing strongly that Notch and not other Fbxw7 substrates are primarily responsible for these phenotypes, the genetic inactivation of Notch pathway components reversed the impaired ISV growth in the zebrafish embryo as well as sprouting and proliferation in the mouse retina. Our findings establish that Fbxw7 is a potent positive regulator of angiogenesis that limits the activity of Notch in the endothelium of the growing vasculature.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , Receptors, Notch/metabolism , Ubiquitin-Protein Ligases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mice , Mice, Transgenic , Morpholinos/pharmacology , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Notch/genetics , Retina/cytology , Retina/metabolism , Ubiquitin-Protein Ligase Complexes , Ubiquitin-Protein Ligases/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL